1
|
Tamalunas A, Wendt A, Springer F, Vigodski V, Trieb M, Eitelberger N, Poth H, Ciotkowska A, Rutz B, Hu S, Schulz H, Ledderose S, Rogenhofer N, Kolben T, Nössner E, Stief CG, Hennenberg M. Immunomodulatory imide drugs inhibit human detrusor smooth muscle contraction and growth of human detrusor smooth muscle cells, and exhibit vaso-regulatory functions. Biomed Pharmacother 2024; 177:117066. [PMID: 38981242 DOI: 10.1016/j.biopha.2024.117066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND The immunomodulatory imide drugs (IMiDs) thalidomide, lenalidomide and pomalidomide may exhibit therapeutic efficacy in the prostate. In lower urinary tract symptoms (LUTS), voiding and storage disorders may arise from benign prostate hyperplasia, or overactive bladder. While current therapeutic options target smooth muscle contraction or cell proliferation, side effects are mostly cardiovascular. Therefore, we investigated effects of IMiDs on human detrusor and porcine artery smooth muscle contraction, and growth-related functions in detrusor smooth muscle cells (HBdSMC). METHODS Cell viability was assessed by CCK8, and apoptosis and cell death by flow cytometry in cultured HBdSMC. Contractions of human detrusor tissues and porcine interlobar and coronary arteries were induced by contractile agonists, or electric field stimulation (EFS) in the presence or absence of an IMID using an organ bath. Proliferation was assessed by EdU assay and colony formation, cytoskeletal organization by phalloidin staining, RESULTS: Depending on tissue type, IMiDs inhibited cholinergic contractions with varying degree, up to 50 %, while non-cholinergic contractions were inhibited up to 80 % and 60 % for U46619 and endothelin-1, respectively, and EFS-induced contractions up to 75 %. IMiDs reduced viable HBdSM cells in a time-dependent manner. Correspondingly, proliferation was reduced, without showing pro-apoptotic effects. In parallel, IMiDs induced cytoskeletal disorganization. CONCLUSIONS IMiDs exhibit regulatory functions in various smooth muscle-rich tissues, and of cell proliferation in the lower urinary tract. This points to a novel drug class effect for IMiDs, in which the molecular mechanisms of action of IMiDs merit further consideration for the application in LUTS.
Collapse
Affiliation(s)
- Alexander Tamalunas
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany; Immunoanalytics Research Group Tissue Control of Immunocytes, Helmholtz Center Munich, Munich, Germany.
| | - Amin Wendt
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Florian Springer
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Victor Vigodski
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Moritz Trieb
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | | | - Henrik Poth
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Anna Ciotkowska
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Beata Rutz
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Sheng Hu
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Heiko Schulz
- Department of Pathology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Stephan Ledderose
- Department of Pathology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Nina Rogenhofer
- Department of Obstetrics and Gynecology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Thomas Kolben
- Department of Obstetrics and Gynecology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Elfriede Nössner
- Immunoanalytics Research Group Tissue Control of Immunocytes, Helmholtz Center Munich, Munich, Germany
| | - Christian G Stief
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Martin Hennenberg
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| |
Collapse
|
2
|
Tang R, Zhou Y, Mei S, Xu Q, Feng J, Xing S, Gao Y, Qin S, He Z. Fibrotic extracellular vesicles contribute to mechanical ventilation-induced pulmonary fibrosis development by activating lung fibroblasts via JNK signalling pathway: an experimental study. BMJ Open Respir Res 2023; 10:e001753. [PMID: 37620111 PMCID: PMC10450055 DOI: 10.1136/bmjresp-2023-001753] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Recent research has revealed that mechanical ventilation (MV) could initiate ventilator-induced lung injury along with the initiation of the process of pulmonary fibrosis (PF), leading to MV-induced PF (MVPF). However, the underlying mechanism remains unclear. This study aimed to explore the role of MV-induced extracellular vesicles (MV-EVs) and the c-Jun N-terminal kinase (JNK) signalling pathway in the pathogenesis of MVPF in vivo and in vitro. The process of MV is accompanied by the secretion of MV-EVs, which could induce lung fibroblast activation. Furthermore, single-cell RNA-sequencing analysis revealed that the JNK pathway in lung fibroblasts was activated after MV initiation. Inhibiting the JNK pathway could both restrain MV-EV-induced lung fibroblast activation in vitro or reduce the severity of MVPF in vivo. In conclusion, this study demonstrated that MV-EVs contribute to MVPF progression by activating lung fibroblasts via the JNK signalling pathway and that inhibiting the secretion of EV and the activation of the JNK signalling pathway is a promising strategy for treating MVPF.
Collapse
Affiliation(s)
- Ri Tang
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Zhou
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuya Mei
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiaoyi Xu
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinhua Feng
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunpeng Xing
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Gao
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaojie Qin
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengyu He
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Dsouza NN, Alampady V, Baby K, Maity S, Byregowda BH, Nayak Y. Thalidomide interaction with inflammation in idiopathic pulmonary fibrosis. Inflammopharmacology 2023; 31:1167-1182. [PMID: 36966238 PMCID: PMC10039777 DOI: 10.1007/s10787-023-01193-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/04/2023] [Indexed: 03/27/2023]
Abstract
The "Thalidomide tragedy" is a landmark in the history of the pharmaceutical industry. Despite limited clinical trials, there is a continuous effort to investigate thalidomide as a drug for cancer and inflammatory diseases such as rheumatoid arthritis, lepromatous leprosy, and COVID-19. This review focuses on the possibilities of targeting inflammation by repurposing thalidomide for the treatment of idiopathic pulmonary fibrosis (IPF). Articles were searched from the Scopus database, sorted, and selected articles were reviewed. The content includes the proven mechanisms of action of thalidomide relevant to IPF. Inflammation, oxidative stress, and epigenetic mechanisms are major pathogenic factors in IPF. Transforming growth factor-β (TGF-β) is the major biomarker of IPF. Thalidomide is an effective anti-inflammatory drug in inhibiting TGF-β, interleukins (IL-6 and IL-1β), and tumour necrosis factor-α (TNF-α). Thalidomide binds cereblon, a process that is involved in the proposed mechanism in specific cancers such as breast cancer, colon cancer, multiple myeloma, and lung cancer. Cereblon is involved in activating AMP-activated protein kinase (AMPK)-TGF-β/Smad signalling, thereby attenuating fibrosis. The past few years have witnessed an improvement in the identification of biomarkers and diagnostic technologies in respiratory diseases, partly because of the COVID-19 pandemic. Hence, investment in clinical trials with a systematic plan can help repurpose thalidomide for pulmonary fibrosis.
Collapse
Affiliation(s)
- Nikitha Naomi Dsouza
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varun Alampady
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishnaprasad Baby
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Swastika Maity
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bharath Harohalli Byregowda
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
4
|
Yiou W, Zhihong W, Shibai Z, Shanni L, Wenwei Q. MiR-326 regulates cell proliferation and apoptosis in fibroblast-like synoviocytes in rheumatoid arthritis. Hum Cell 2023; 36:987-996. [PMID: 36749538 DOI: 10.1007/s13577-023-00873-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 01/28/2023] [Indexed: 02/08/2023]
Abstract
The dysregulation of microRNAs plays a critical role in the development of rheumatoid arthritis (RA). This study aims to explore the functional significance of miR-326 in RA. The RT-qPCR results showed that miR-326 was downregulated in synovial tissues of RA patients and RA fibroblast-like synoviocytes (RA-FLS). We found that miR-326 could target and reduce the expression of inhibitor of DNA binding 1 (Id1). MTT assay and flow cytometry were conducted to explore the biological function of miR-326. Our data revealed that the upregulation of miR-326 suppressed cell proliferation and induced apoptosis in RA-FLS. In collagen-induced arthritis mice, intraarticular injection of lentivirus carrying miR-326 overexpression vectors could reduce the arthritis score and attenuate synovial inflammation and cartilage destruction. We also found that long non-coding RNA-Ewing sarcoma-associated transcript 1 (lncRNA-EWSAT1) was significantly increased in RA synovial tissues and RA-FLS. The RNA immunoprecipitation and RNA pull-down assay indicated that lncRNA-EWSAT1 directly bound and negatively regulated the expression of miR-326. Knockdown of lncRNA-EWSAT1 could upregulate miR-326 expression and attenuate its proliferation inhibition and apoptosis induction effect in RA-FLS. In conclusion, the lncRNA-EWSAT1/miR-326 axis might provide a novel therapeutic target in the treatment of RA.
Collapse
Affiliation(s)
- Wang Yiou
- Department of Plastic and Reconstructive Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Wang Zhihong
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei Province, China
| | - Zhu Shibai
- Department of Orthopedic Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100176, China
| | - Li Shanni
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qian Wenwei
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
5
|
Estornut C, Milara J, Bayarri MA, Belhadj N, Cortijo J. Targeting Oxidative Stress as a Therapeutic Approach for Idiopathic Pulmonary Fibrosis. Front Pharmacol 2022; 12:794997. [PMID: 35126133 PMCID: PMC8815729 DOI: 10.3389/fphar.2021.794997] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/10/2021] [Indexed: 01/19/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease characterized by an abnormal reepithelialisation, an excessive tissue remodelling and a progressive fibrosis within the alveolar wall that are not due to infection or cancer. Oxidative stress has been proposed as a key molecular process in pulmonary fibrosis development and different components of the redox system are altered in the cellular actors participating in lung fibrosis. To this respect, several activators of the antioxidant machinery and inhibitors of the oxidant species and pathways have been assayed in preclinical in vitro and in vivo models and in different clinical trials. This review discusses the role of oxidative stress in the development and progression of IPF and its underlying mechanisms as well as the evidence of oxidative stress in human IPF. Finally, we analyze the mechanism of action, the efficacy and the current status of different drugs developed to inhibit the oxidative stress as anti-fibrotic therapy in IPF.
Collapse
Affiliation(s)
- Cristina Estornut
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- *Correspondence: Cristina Estornut, ; Javier Milara,
| | - Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
- *Correspondence: Cristina Estornut, ; Javier Milara,
| | - María Amparo Bayarri
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Nada Belhadj
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
- Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
6
|
Tamalunas A, Sauckel C, Ciotkowska A, Rutz B, Wang R, Huang R, Li B, Stief CG, Gratzke C, Hennenberg M. Lenalidomide and pomalidomide inhibit growth of prostate stromal cells and human prostate smooth muscle contraction. Life Sci 2021; 281:119771. [PMID: 34186051 DOI: 10.1016/j.lfs.2021.119771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022]
Abstract
AIMS Medical treatment for lower urinary tract symptoms secondary to benign prostatic hyperplasia is characterized by an unfavorable balance between limited efficacy and pronounced side effects. We recently reported, that thalidomide reduces prostate smooth muscle contraction and inhibits cell growth. Like thalidomide, its analogs lenalidomide and pomalidomide are also in clinical use. Therefore, we investigated the effects of lenalidomide and pomalidomide on human prostate smooth muscle contraction, cytoskeletal organization, and growth-related functions in stromal cells. MATERIALS AND METHODS Proliferation was assessed by EdU assay and colony formation, cytoskeletal organization by phalloidin staining, cell viability by CCK8, and apoptosis and cell death by flow cytometry in cultured prostate stromal cells (WPMY-1). Contractions of human prostate tissues from radical prostatectomy were induced by methoxamine, noradrenaline, phenylephrine, endothelin-1, U46619, or electric field stimulation (EFS) in an organ bath. KEY FINDINGS Proliferation of WPMY-1 cells was significantly reduced by lenalidomide (5-200 μM) and pomalidomide (2.5-5 μM). In parallel, organization of actin filaments collapsed after treatment with lenalidomide and pomalidomide. Lenalidomide and pomalidomide inhibited both adrenergic contractions and non-adrenergic contractions as well as neurogenic contractions induced by EFS. Neither reduction in viability, nor increase in cell death or apoptosis was observed in WPMY-1 cells. SIGNIFICANCE Thalidomide-derivatives impair growth of human prostate stromal cells, without showing a decrease in cell viability and, in parallel, inhibit adrenergic, neurogenic, and non-adrenergic contractions by breakdown of the actin cytoskeleton. Urodynamic effects in vivo appear possible.
Collapse
Affiliation(s)
| | - Cora Sauckel
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Anna Ciotkowska
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Beata Rutz
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Ruixiao Wang
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Ru Huang
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Bingsheng Li
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Christian G Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | | | - Martin Hennenberg
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Tamalunas A, Sauckel C, Ciotkowska A, Rutz B, Wang R, Huang R, Li B, Stief CG, Gratzke C, Hennenberg M. Inhibition of human prostate stromal cell growth and smooth muscle contraction by thalidomide: A novel remedy in LUTS? Prostate 2021; 81:377-389. [PMID: 33687083 DOI: 10.1002/pros.24114] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/19/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Medical treatment in benign prostatic hyperplasia targets prostate size to prevent disease progression, complications, and surgery, and prostate smooth muscle tone for rapid relief of lower urinary tract symptoms. Combination therapies are still required to target both at once. However, current medications are insufficient, due to an unfavorable balance between side effects and efficacy. The limited efficacy of α1 -blockers may be due to nonadrenergic mediators like endothelin-1 and thromboxane A2 (TXA2 ), which keep up prostate smooth muscle contraction even in the presence of α1 -blockers. Consequently, future options with higher efficacy need to target α1 -adrenergic and nonadrenergic contractions as well as stromal cell growth at once. Thalidomide has been approved as an oral medication for various diseases, including the treatment of prostate cancer. Therefore, we investigated the effect of thalidomide on cellular functions of prostate stromal cells and human prostate smooth muscle contraction. METHODS Cytoskeletal organization was visualized by phalloidin staining, cell growth was assessed by 5-ethynyl-2'-deoxyuridine assay, cell viability by cell counting kit-8, and apoptosis and cell death by flow cytometry in cultured prostate stromal cells (WPMY-1). Contractions of human prostate tissues from radical prostatectomy were studied in an organ bath, where they were induced by the α1 -adrenoceptor agonists methoxamine, noradrenaline, phenylephrine, and the nonadrenergic agonists endothelin-1, TXA2 analog U46619, or electric field stimulation (EFS). RESULTS Thalidomide significantly reduced the proliferation of WPMY-1 cells, which was time- and concentration-dependent (10-300 µM). In parallel, organization of actin filaments collapsed after treatment with thalidomide. Thalidomide (30-100 µM) inhibited noradrenaline-, phenylephrine-, and methoxamine-induced contractions, as well as nonadrenergic contractions induced by endothelin-1 and U46619, and neurogenic contractions induced by EFS. No reduction in viability and no increases in apoptosis or in cell death were observed in WPMY-1 cells. CONCLUSIONS Thalidomide impairs the growth of human prostate stromal cells, without showing a decrease in cell viability. In parallel, thalidomide inhibits adrenergic, neurogenic, and nonadrenergic contractions. This may be explained by a breakdown of the actin cytoskeleton. In vivo, urodynamic effects of thalidomide appear possible and may even exceed those of α1 -blockers or combination therapies.
Collapse
Affiliation(s)
| | - Cora Sauckel
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Anna Ciotkowska
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Beata Rutz
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Ruixiao Wang
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Ru Huang
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Bingsheng Li
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Christian G Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | | | - Martin Hennenberg
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
8
|
Finato AC, Almeida DF, Dos Santos AR, Nascimento DC, Cavalcante RS, Mendes RP, Soares CT, Paniago AMM, Venturini J. Evaluation of antifibrotic and antifungal combined therapies in experimental pulmonary paracoccidioidomycosis. Med Mycol 2020; 58:667-678. [PMID: 31578565 DOI: 10.1093/mmy/myz100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/27/2019] [Accepted: 09/11/2019] [Indexed: 01/04/2023] Open
Abstract
Paracoccidioidomycosis (PCM) is a systemic mycosis caused by the Paracoccidioides genus. Most of the patients with chronic form present sequelae, like pulmonary fibrosis, with no effective treatment, leading to impaired lung functions. In the present study, we aimed to investigate the antifibrotic activity of three compounds: pentoxifylline (PTX), azithromycin (AZT), and thalidomide (Thal) in a murine model of pulmonary PCM treated with itraconazole (ITC) or cotrimoxazole (CMX). BALB/c mice were inoculated with P. brasiliensis (Pb) by the intratracheal route and after 8 weeks, they were submitted to one of the following six treatments: PTX/ITC, PTX/CMX, AZT/ITC, AZT/CMX, Thal/ITC, and Thal/CMX. After 8 weeks of treatment, the lungs were collected for determination of fungal burden, production of OH-proline, deposition of reticulin fibers, and pulmonary concentrations of cytokines and growth factors. Pb-infected mice treated with PTX/ITC presented a reduction in the pulmonary concentrations of OH-proline, associated with lower concentrations of interleukin (IL)-6, IL-17, and transforming growth factor (TGF)-β1 and higher concentrations of IL-10 compared to the controls. The Pb-infected mice treated with AZT/CMX exhibited decreased pulmonary concentrations of OH-proline associated with lower levels of TGF-β1, and higher levels of IL-10 compared controls. The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-γ and VEGF, and decreased concentrations of IL-6, IL-1β, IL-17, and TGF-β1. In conclusion, our findings reinforce the antifibrotic role of PTX only when associated with ITC, and AZT only when associated with CMX, but Thal did not show any action upon addition.
Collapse
Affiliation(s)
- Angela C Finato
- Faculdade de Ciências. Universidade Estadual Paulista (UNESP), 17033-360 Bauru, SP, Brazil
| | - Débora F Almeida
- Faculdade de Ciências. Universidade Estadual Paulista (UNESP), 17033-360 Bauru, SP, Brazil.,Faculdade de Medicina (FAMED). Universidade Federal do Mato Grosso do Sul (UFMS). 79070-900 Campo Grande, MS, Brazil
| | - Amanda R Dos Santos
- Faculdade de Ciências. Universidade Estadual Paulista (UNESP), 17033-360 Bauru, SP, Brazil.,Faculdade de Medicina (FAMED). Universidade Federal do Mato Grosso do Sul (UFMS). 79070-900 Campo Grande, MS, Brazil
| | | | - Ricardo S Cavalcante
- Faculdade de Medicina de Botucatu. Universidade Estadual Paulista (UNESP), 18618-687 Botucatu, SP, Brazil
| | - Rinaldo P Mendes
- Faculdade de Medicina (FAMED). Universidade Federal do Mato Grosso do Sul (UFMS). 79070-900 Campo Grande, MS, Brazil.,Faculdade de Medicina de Botucatu. Universidade Estadual Paulista (UNESP), 18618-687 Botucatu, SP, Brazil
| | | | - Anamaria M M Paniago
- Faculdade de Medicina (FAMED). Universidade Federal do Mato Grosso do Sul (UFMS). 79070-900 Campo Grande, MS, Brazil
| | - James Venturini
- Faculdade de Medicina (FAMED). Universidade Federal do Mato Grosso do Sul (UFMS). 79070-900 Campo Grande, MS, Brazil
| |
Collapse
|
9
|
Dong X, Li X, Li M, Chen M, Fan Q, Wei W. Antiinflammation and Antioxidant Effects of Thalidomide on Pulmonary Fibrosis in Mice and Human Lung Fibroblasts. Inflammation 2018; 40:1836-1846. [PMID: 28730510 DOI: 10.1007/s10753-017-0625-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In this study, the potential effects of thalidomide (Thal) on bleomycin (BLM)-induced pulmonary fibrosis were investigated. BALB/C mice model of pulmonary fibrosis induced by an intratracheal instillation of BLM was adopted, and then was intraperitoneally injected with Thal (10, 20, 50 mg/kg) daily for 8 days, while the control and BLM-treated mouse groups were injected with a saline solution. The effects of Thal on pulmonary injury were evaluated by the lung wet/dry weight ratios and histopathological examination. Inflammation of lung tissues was assessed by measuring the levels of interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-β in bronchoalveolar lavage fluid. Oxidative stress was evaluated by detecting the levels of reactive oxygen species (ROS), superoxide dismutase (SOD), total antioxidant capacity (T-AOC), and malondialdehyde (MDA) in lung tissue. The results indicated that Thal treatment remarkably attenuated pulmonary fibrosis, oxidative stress, and inflammation in mouse lungs. The antiinflammatory and antioxidant effects of Thal were also found in human lung fibroblasts. Thal administration significantly enhanced the activity of thioredoxin reductase; however, the other enzymes or proteins involved in biologic oxidation-reduction equilibrium were not affected. Our findings indicate that Thal-mediated suppression of pulmonary fibrosis is related to the inhibition of oxidative stress and inflammatory response. In summary, these results may provide a rationale to explore clinical application of Thal for the prevention of pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiaoying Dong
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Xin Li
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Minghui Li
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Ming Chen
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Qian Fan
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Wei Wei
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
10
|
Thalidomide Inhibits TGF-β1-induced Epithelial to Mesenchymal Transition in Alveolar Epithelial Cells via Smad-Dependent and Smad-Independent Signaling Pathways. Sci Rep 2017; 7:14727. [PMID: 29116196 PMCID: PMC5677010 DOI: 10.1038/s41598-017-15239-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/24/2017] [Indexed: 12/18/2022] Open
Abstract
Recent evidence indicates that the epithelial to mesenchymal transition (EMT) in primary alveolar cells (AECs) plays an important role in idiopathic pulmonary fibrosis (IPF). In vivo models have suggested that thalidomide (THL) has anti-fibrotic effects against pulmonary fibrosis, but the underlying mechanism of this effect is not clear. This study investigated whether THL regulates alveolar EMT and the possible mechanisms underlying this process. CCL-149 cells were treated with TGF-β1 in the presence of THL at the indicated concentrations. EMT was assessed by changes in cell morphology and in phenotypic markers. Signaling pathways involved in EMT were characterized by western blot analysis. THL inhibited the TGF-β1 induction of α-SMA, vimentin, MMP-2/-9 and collagen type IV expression and restored the morphological changes in primary alveolar epithelial cells caused by TGF-β1. TGF-β1 induction of α-SMA expression was partially dependent on the activation of p38, JNK, ERK, Akt, Smad 2 and Smad3. Moreover, THL inhibited TGF-β1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3β, Smad 2 and Smad3 without altering the total expression levels of those proteins. These findings indicate that TGF-β1-induced EMT in alveolar epithelial cells is inhibited by THL via both Smad-dependent and non-Smad-dependent signaling pathways and suggests therapeutic approaches for targeting this process in pulmonary fibrosis.
Collapse
|
11
|
Dong X, Li X, Li M, Chen M, Fan Q, Wei W. Inhibitory effects of thalidomide on bleomycin-induced pulmonary fibrosis in rats via regulation of thioredoxin reductase and inflammations. Am J Transl Res 2017; 9:4390-4401. [PMID: 29118902 PMCID: PMC5666049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/06/2017] [Indexed: 06/07/2023]
Abstract
In this study, the potential clinical effects of thalidomide on bleomycin-induced pulmonary fibrosis were investigated. A Sprague-Dawley rats' model of pulmonary fibrosis induced by an intratracheal instillation of bleomycin was adopted. The rats in thalidomide treated groups were intraperitoneally injected with thalidomide (10, 20, 50 mg/kg) daily for 28 days, while the rats in control and bleomycin treated groups were injected with a saline solution. The effects of thalidomide on pulmonary injury were evaluated by the lung wet/dry weight ratios, cell counts, and histopathological examination. Inflammation of lung tissues was assessed by measuring the levels of interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-β in bronchoalveolar lavage fluid (BALF). Oxidative stress was evaluated by detecting the levels of reactive oxygen species (ROS), superoxide dismutase (SOD), total antioxidant capacity (T-AOC), and malondialdehyde (MDA) in lung tissue. The results indicated that thalidomide treatment remarkably attenuated bleomycin-induced pulmonary fibrosis, oxidative stress and inflammation in rats' lung. The anti-inflammatory and anti-oxidative effects of thalidomide were also found in human lung fibroblasts. Thalidomide administration significantly stimulated the activity of thioredoxin reductase, while other enzymes or proteins involved in biologic oxidation-reduction equilibrium were not affected. Our findings indicate that thalidomide-mediated suppression of fibro-proliferation may contribute to the anti-fibrotic effect against bleomycin-induced pulmonary fibrosis. The mechanisms are related to the inhibition of oxidative stress and inflammatory response. In summary, these results may provide a rationale to explore clinical application of thalidomide for the prevention of pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiaoying Dong
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical UniversityTianjin 300052, China
| | - Xin Li
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical UniversityTianjin 300052, China
| | - Minghui Li
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical UniversityTianjin 300052, China
| | - Ming Chen
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical UniversityTianjin 300052, China
| | - Qian Fan
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical UniversityTianjin 300052, China
| | - Wei Wei
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical UniversityTianjin 300052, China
| |
Collapse
|
12
|
Eurlings IMJ, Reynaert NL, van de Wetering C, Aesif SW, Mercken EM, de Cabo R, van der Velden JL, Janssen-Heininger YM, Wouters EFM, Dentener MA. Involvement of c-Jun N-Terminal Kinase in TNF-α-Driven Remodeling. Am J Respir Cell Mol Biol 2017; 56:393-401. [PMID: 27875656 DOI: 10.1165/rcmb.2015-0195oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lung tissue remodeling in chronic obstructive pulmonary disease (COPD) is characterized by airway wall thickening and/or emphysema. Although the bronchial and alveolar compartments are functionally independent entities, we recently showed comparable alterations in matrix composition comprised of decreased elastin content and increased collagen and hyaluronan contents of alveolar and small airway walls. Out of several animal models tested, surfactant protein C (SPC)-TNF-α mice showed remodeling in alveolar and airway walls similar to what we observed in patients with COPD. Epithelial cells are able to undergo a phenotypic shift, gaining mesenchymal properties, a process in which c-Jun N-terminal kinase (JNK) signaling is involved. Therefore, we hypothesized that TNF-α induces JNK-dependent epithelial plasticity, which contributes to lung matrix remodeling. To this end, the ability of TNF-α to induce a phenotypic shift was assessed in A549, BEAS2B, and primary bronchial epithelial cells, and phenotypic markers were studied in SPC-TNF-α mice. Phenotypic markers of mesenchymal cells were elevated both in vitro and in vivo, as shown by the expression of vimentin, plasminogen activator inhibitor-1, collagen, and matrix metalloproteinases. Concurrently, the expression of the epithelial markers, E-cadherin and keratin 7 and 18, was attenuated. A pharmacological inhibitor of JNK attenuated this phenotypic shift in vitro, demonstrating involvement of JNK signaling in this process. Interestingly, activation of JNK signaling was also clearly present in lungs of SPC-TNF-α mice and patients with COPD. Together, these data show a role for TNF-α in the induction of a phenotypic shift in vitro, resulting in increased collagen production and the expression of elastin-degrading matrix metalloproteinases, and provide evidence for involvement of the TNF-α-JNK axis in extracellular matrix remodeling.
Collapse
Affiliation(s)
- Irene M J Eurlings
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Niki L Reynaert
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Cheryl van de Wetering
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Scott W Aesif
- 2 Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Evi M Mercken
- 3 Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland; and
| | - Rafael de Cabo
- 3 Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland; and
| | - Jos L van der Velden
- 4 Department of Pathology, University of Vermont College of Medicine, Burlington, Vermont
| | | | - Emiel F M Wouters
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Mieke A Dentener
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
13
|
Salvianolic Acid B Attenuates Experimental Pulmonary Fibrosis through Inhibition of the TGF-β Signaling Pathway. Sci Rep 2016; 6:27610. [PMID: 27278104 PMCID: PMC4899783 DOI: 10.1038/srep27610] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 05/20/2016] [Indexed: 12/22/2022] Open
Abstract
Pulmonary fibrosis is a progressive and fatal disorder. In our previous study, we found that the Yiqihuoxue formula (YQHX), a prescription of Traditional Chinese Medicine, had a curative effect on scleroderma, a typical fibrotic disease. The aim of this study was to determine the key ingredient mediating the therapeutic effects of YQHX and to examine its effect on pulmonary fibrosis, including its mechanism. Luciferase reporter assays showed that the most important anti-fibrotic component of the YQHX was Salviae miltiorrhiza (SM). Experiments performed using a bleomycin-instilled mouse model of pulmonary fibrosis showed that Salvianolic acid B (SAB), the major ingredient of SM, had strong anti-inflammatory and anti-fibrotic effects through its inhibition of inflammatory cell infiltration, alveolar structure disruption, and collagen deposition. Furthermore, SAB suppressed TGF-β-induced myofibroblastic differentiation of MRC-5 fibroblasts and TGF-β-mediated epithelial-to-mesenchymal transition of A549 cells by inhibiting both Smad-dependent signaling and the Smad-independent MAPK pathway. Taken together, our results suggest that SM is the key anti-fibrotic component of the YQHX and that SAB, the major ingredient of SM, alleviates experimental pulmonary fibrosis both in vivo and in vitro by inhibiting the TGF-β signaling pathway. Together, these results suggest that SAB potently inhibits pulmonary fibrosis.
Collapse
|
14
|
Zhou MI, Chen DL, Jiang T, Feng YM, Han XL. Effects of bone marrow-derived mesenchymal stem cells transfected with survivin on pulmonary fibrosis in mice. Exp Ther Med 2015; 10:1857-1864. [PMID: 26640562 DOI: 10.3892/etm.2015.2715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 07/17/2015] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to investigate the effects of bone marrow-derived mesenchymal stem cells (BMSCs) transfected with survivin on lung fibrosis in mice. Mice with bleomycin-induced pulmonary fibrosis were allocated at random to group A, B or C, and injected with 1×106 survivin gene-expressing BMSCs, 1×106 BMSCs or normal saline, respectively. A total of 6 mice were sacrificed from each group on days 7, 14 and 28 after treatment. The extent of alveolitis and pulmonary fibrosis was assessed and the apoptotic rates of the BMSCs and survivin-expressing BMSCs were detected. The content of surfactant protein A (SP-A) in the lung and hydroxyproline (Hyp) in the serum was measured. The mRNA expression levels of transforming growth factor (TGF)-β1 and matrix metalloproteinase (MMP)-9 in the lung tissue of the mice was detected. Furthermore, the protein expression levels of caspase-3 and -9 were detected. The apoptotic rates of the BMSCs (group B) and survivin-expressing BMSCs (group A) were 14.466±1.953 and 7.718±0.493%, respectively. The degree of lung fibrosis in groups A and B was reduced compared with that in group C. The hydroxyproline content in groups A and B was reduced compared with that in group C, and the SP-A content in groups A and B was increased compared with that in group C. The mRNA expression levels of TGF-β1 in group A were reduced compared with those in group B, and the levels in group B were reduced compared with those in group C. By contrast, the mRNA expression levels of MMP-9 in group A were increased compared with those in groups B and C, and the levels in group B were increased compared with those in group A. The expression levels of caspase-3 and -9 in group A were elevated compared with those in groups B and C. In conclusion, BMSCs are effective in preventing bleomycin-induced lung fibrosis, and survivin may enhance the protective effects of BMSCs.
Collapse
Affiliation(s)
- M I Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Dong-Ling Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Tao Jiang
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Yan-Mei Feng
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Xiao-Li Han
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| |
Collapse
|
15
|
De Paepe B, Zschüntzsch J. Scanning for Therapeutic Targets within the Cytokine Network of Idiopathic Inflammatory Myopathies. Int J Mol Sci 2015; 16:18683-713. [PMID: 26270565 PMCID: PMC4581266 DOI: 10.3390/ijms160818683] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/13/2015] [Accepted: 07/15/2015] [Indexed: 12/17/2022] Open
Abstract
The idiopathic inflammatory myopathies (IIM) constitute a heterogeneous group of chronic disorders that include dermatomyositis (DM), polymyositis (PM), sporadic inclusion body myositis (IBM) and necrotizing autoimmune myopathy (NAM). They represent distinct pathological entities that, most often, share predominant inflammation in muscle tissue. Many of the immunopathogenic processes behind the IIM remain poorly understood, but the crucial role of cytokines as essential regulators of the intramuscular build-up of inflammation is undisputed. This review describes the extensive cytokine network within IIM muscle, characterized by strong expression of Tumor Necrosis Factors (TNFα, LTβ, BAFF), Interferons (IFNα/β/γ), Interleukins (IL-1/6/12/15/18/23) and Chemokines (CXCL9/10/11/13, CCL2/3/4/8/19/21). Current therapeutic strategies and the exploration of potential disease modifying agents based on manipulation of the cytokine network are provided. Reported responses to anti-TNFα treatment in IIM are conflicting and new onset DM/PM has been described after administration of anti-TNFα agents to treat other diseases, pointing to the complex effects of TNFα neutralization. Treatment with anti-IFNα has been shown to suppress the IFN type 1 gene signature in DM/PM patients and improve muscle strength. Beneficial effects of anti-IL-1 and anti-IL-6 therapy have also been reported. Cytokine profiling in IIM aids the development of therapeutic strategies and provides approaches to subtype patients for treatment outcome prediction.
Collapse
Affiliation(s)
- Boel De Paepe
- Neuromuscular Reference Center, Laboratory for Neuropathology, 10K12E, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Centre, Göttingen University, 37075 Göttingen, Germany.
| |
Collapse
|
16
|
Biochemical and histological impact of direct renin inhibition by aliskiren on myofibroblasts activation and differentiation in bleomycin induced pulmonary fibrosis in adult mice. Tissue Cell 2015; 47:373-81. [DOI: 10.1016/j.tice.2015.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 05/01/2015] [Accepted: 05/02/2015] [Indexed: 01/25/2023]
|